Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Circ Res ; 128(1): 62-75, 2021 01 08.
Article in English | MEDLINE | ID: mdl-33070717

ABSTRACT

RATIONALE: Hemorrhagic complications represent a major limitation of intravenous thrombolysis using tPA (tissue-type plasminogen activator) in patients with ischemic stroke. The expression of tPA receptors on immune cells raises the question of what effects tPA exerts on these cells and whether these effects contribute to thrombolysis-related hemorrhagic transformation. OBJECTIVE: We aim to determine the impact of tPA on immune cells and investigate the association between observed immune alteration with hemorrhagic transformation in ischemic stroke patients and in a rat model of embolic stroke. METHODS AND RESULTS: Paired blood samples were collected before and 1 hour after tPA infusion from 71 patients with ischemic stroke. Control blood samples were collected from 27 ischemic stroke patients without tPA treatment. A rat embolic middle cerebral artery occlusion model was adopted to investigate the underlying mechanisms of hemorrhagic transformation. We report that tPA induces a swift surge of circulating neutrophils and T cells with profoundly altered molecular features in ischemic stroke patients and a rat model of focal embolic stroke. tPA exacerbates endothelial injury, increases adhesion and migration of neutrophils and T cells, which are associated with brain hemorrhage in rats subjected to embolic stroke. Genetic ablation of annexin A2 in neutrophils and T cells diminishes the effect of tPA on these cells. Decoupling the interaction between mobilized neutrophils/T cells and the neurovascular unit, achieved via a S1PR (sphingosine-1-phosphate receptor) 1 modulator RP101075 and a CCL2 (C-C motif chemokine ligand 2) synthesis inhibitor bindarit, which block lymphocyte egress and myeloid cell recruitment, respectively, attenuates hemorrhagic transformation and improves neurological function after tPA thrombolysis. CONCLUSIONS: Our findings suggest that immune invasion of the neurovascular unit represents a previously unrecognized mechanism underlying tPA-mediated brain hemorrhage, which can be overcome by precise immune modulation during thrombolytic therapy.


Subject(s)
Embolic Stroke/drug therapy , Fibrinolytic Agents/toxicity , Infarction, Middle Cerebral Artery/drug therapy , Intracranial Hemorrhages/chemically induced , Ischemic Stroke/drug therapy , Neutrophils/drug effects , T-Lymphocytes/drug effects , Thrombolytic Therapy , Tissue Plasminogen Activator/toxicity , Animals , Annexin A2/metabolism , Cell Line , Chemokine CCL2/metabolism , Chemotaxis, Leukocyte/drug effects , Disease Models, Animal , Embolic Stroke/blood , Embolic Stroke/immunology , Female , Fibrinolytic Agents/administration & dosage , Humans , Infarction, Middle Cerebral Artery/blood , Infarction, Middle Cerebral Artery/immunology , Infusions, Intravenous , Intracranial Hemorrhages/blood , Intracranial Hemorrhages/immunology , Ischemic Stroke/blood , Ischemic Stroke/immunology , Male , Neutrophil Infiltration/drug effects , Neutrophils/immunology , Neutrophils/metabolism , Rats, Wistar , Sphingosine-1-Phosphate Receptors/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tissue Plasminogen Activator/administration & dosage
2.
Nucleic Acid Ther ; 30(2): 94-103, 2020 04.
Article in English | MEDLINE | ID: mdl-32043907

ABSTRACT

Inotersen, a 2'-O-methoxyethyl (2'-MOE) phosphorothioate antisense oligonucleotide, reduced disease progression and improved quality of life in patients with hereditary transthyretin amyloidosis with polyneuropathy (hATTR-PN) in the NEURO-TTR and NEURO-TTR open-label extension (OLE) trials. However, 300 mg/week inotersen treatment was associated with platelet count reductions in several patients. Mean platelet counts in patients in the NEURO-TTR-inotersen group remained ≥140 × 109/L in 50% and ≥100 × 109/L in 80% of the subjects. However, grade 4 thrombocytopenia (<25 × 109/L) occurred in three subjects in NEURO-TTR trial, and one of these suffered a fatal intracranial hemorrhage. The two others were treated successfully with corticosteroids and discontinuation of inotersen. Investigations in a subset of subjects in NEURO-TTR (n = 17 placebo; n = 31 inotersen) and OLE (n = 33) trials ruled out direct myelotoxicity, consumptive coagulopathy, and heparin-induced thrombocytopenia. Antiplatelet immunoglobulin G (IgG) antibodies were detected at baseline in 5 of 31 (16%) inotersen-treated subjects in NEURO-TTR, 4 of whom eventually developed grade 1 or 2 thrombocytopenia while on the drug. In addition, 24 subjects in the same group developed treatment-emergent antiplatelet IgG antibodies, of which 2 developed grade 2, and 3 developed grade 4 thrombocytopenia. Antiplatelet IgG antibodies in two of the three grade 4 thrombocytopenia subjects targeted GPIIb/IIIa. Plasma cytokines previously implicated in immune dysregulation, such as interleukin (IL)-23 and a proliferation-inducing ligand (APRIL) were often above the normal range at baseline. Collectively, these findings suggest an underlying immunologic dysregulation predisposing some individuals to immune-mediated thrombocytopenia during inotersen treatment.


Subject(s)
Amyloid Neuropathies, Familial/drug therapy , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides/administration & dosage , Thrombocytopenia/blood , Adult , Aged , Amyloid Neuropathies, Familial/genetics , Amyloid Neuropathies, Familial/immunology , Amyloid Neuropathies, Familial/pathology , Female , Genetic Predisposition to Disease , Humans , Immune System Diseases/chemically induced , Immune System Diseases/immunology , Immune System Diseases/pathology , Immunoglobulin G , Intracranial Hemorrhages/chemically induced , Intracranial Hemorrhages/immunology , Intracranial Hemorrhages/pathology , Male , Middle Aged , Oligodeoxyribonucleotides, Antisense/administration & dosage , Oligonucleotides/adverse effects , Oligonucleotides, Antisense/adverse effects , Quality of Life , Thrombocytopenia/chemically induced , Thrombocytopenia/immunology , Thrombocytopenia/pathology
3.
Neuroreport ; 29(15): 1239-1243, 2018 10 17.
Article in English | MEDLINE | ID: mdl-30096131

ABSTRACT

Hemorrhagic stroke is a major risk factor for cognitive impairment. Our study aimed to measure the effect of ginkgo biloba extract (EGB761) on the cognitive ability and inflammatory expression in hemorrhagic stroke model SD rats and to analyze their relationship. Forty SD rats were divided randomly into an SD group (normal control SD rats), an SD+EGB761 group (normal control SD rats supplemented with 45 mg/kg EGB761), a CO group (hemorrhagic stroke model SD rats using collagenase), and a CO+EGB761 group (hemorrhagic stroke model SD rats supplemented with 45 mg/kg EGB761) consisting of 10 rats, respectively. The Y-electric maze test was selected to measure the cognitive function in four groups. Furthermore, enzyme-linked immunosorbent assay and real-time PCR were, respectively, applied for detecting the protein and gene expression profiles of inflammatory factors in primary cultured microglia. Compared with rats in the SD group, the average time of electrical simulation for mastering criteria was prolonged in the CO group (P<0.05). Furthermore, expression levels of proinflammatory cytokines interleukin-1ß (IL-1ß), IL-6, and tumor necrosis factor-α and anti-inflammatory cytokines IL-4, IL-10, and tumor necrosis factor-ß were significantly increased and decreased, respectively, in rats of the CO group compared with the SD group (P<0.05). The results of electrical simulation time, inflammatory factors protein, and gene expression profile in rats of the CO+EGB761 group compared with the CO group were opposite to above contrast (P<0.05). Ginkgo biloba extract could alleviate the cognitive dysfunction after hemorrhagic stroke in SD rats; this is associated with regulating the expression of inflammatory factors secreted by microglia.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cognition/drug effects , Intracranial Hemorrhages/drug therapy , Nootropic Agents/pharmacology , Plant Extracts/pharmacology , Stroke/drug therapy , Animals , Cells, Cultured , Cognition/physiology , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/immunology , Cognitive Dysfunction/pathology , Disease Models, Animal , Gene Expression/drug effects , Ginkgo biloba , Intracranial Hemorrhages/immunology , Intracranial Hemorrhages/pathology , Intracranial Hemorrhages/psychology , Male , Maze Learning/drug effects , Microglia/drug effects , Microglia/immunology , Microglia/pathology , Random Allocation , Rats, Sprague-Dawley , Stroke/immunology , Stroke/pathology , Stroke/psychology
4.
Arterioscler Thromb Vasc Biol ; 38(8): 1761-1771, 2018 08.
Article in English | MEDLINE | ID: mdl-29903733

ABSTRACT

Objective- Hemorrhagic transformation is a serious complication of ischemic stroke after recanalization therapies. This study aims to identify mechanisms underlying hemorrhagic transformation after cerebral ischemia/reperfusion. Approach and Results- We used wild-type mice and Selplg-/- and Fut7-/- mice defective in P-selectin binding and lymphopenic Rag2-/- mice. We induced 30-minute or 45-minute ischemia by intraluminal occlusion of the middle cerebral artery and assessed hemorrhagic transformation at 48 hours with a hemorrhage grading score, histological means, brain hemoglobin content, or magnetic resonance imaging. We depleted platelets and adoptively transferred T cells of the different genotypes to lymphopenic mice. Interactions of T cells with platelets in blood were studied by flow cytometry and image stream technology. We show that platelet depletion increased the bleeding risk only after large infarcts. Lymphopenia predisposed to hemorrhagic transformation after severe stroke, and adoptive transfer of T cells prevented hemorrhagic transformation in lymphopenic mice. CD4+ memory T cells were the subset of T cells binding P-selectin and platelets through functional P-selectin glycoprotein ligand-1. Mice defective in P-selectin binding had a higher hemorrhagic score than wild-type mice. Adoptive transfer of T cells defective in P-selectin binding into lymphopenic mice did not prevent hemorrhagic transformation. Conclusions- The study identifies lymphopenia as a previously unrecognized risk factor for secondary hemorrhagic transformation in mice after severe ischemic stroke. T cells prevent hemorrhagic transformation by their capacity to bind platelets through P-selectin. The results highlight the role of T cells in bridging immunity and hemostasis in ischemic stroke.


Subject(s)
Adoptive Transfer , Blood Platelets/metabolism , CD4-Positive T-Lymphocytes/transplantation , Infarction, Middle Cerebral Artery/therapy , Intracranial Hemorrhages/prevention & control , Lymphopenia/therapy , P-Selectin/metabolism , Reperfusion Injury/prevention & control , Reperfusion/adverse effects , Animals , Blood Platelets/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , Fucosyltransferases/genetics , Fucosyltransferases/metabolism , Genotype , Immunologic Memory , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/immunology , Infarction, Middle Cerebral Artery/metabolism , Intracranial Hemorrhages/genetics , Intracranial Hemorrhages/immunology , Intracranial Hemorrhages/metabolism , Lymphopenia/genetics , Lymphopenia/immunology , Lymphopenia/metabolism , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , P-Selectin/immunology , Reperfusion Injury/genetics , Reperfusion Injury/immunology , Reperfusion Injury/metabolism , Risk Factors , Time Factors
5.
Brain Res Bull ; 127: 226-233, 2016 10.
Article in English | MEDLINE | ID: mdl-27746369

ABSTRACT

Inflammation and disruption of the blood-brain barrier (BBB) cause oedema and secondary brain injury after intracranial haemorrhage (ICH), which is closely related to patient prognosis. Methylprednisolone sodium succinate (MPSS), a well-known immunosuppressive agent, is widely applied in many diseases to inhibit inflammation. In this study, we investigated the effect of MPSS on inflammation and disruption of the BBB in a model mouse of ICH. ICH was induced by injecting collagenase into the right striatum of male C57/BL mice. Permeability of BBB was measured with Evans Blue assay and brain oedema was detected by measurement of brain water content. Expressions of NF-κB, TLR4, occludin, ZO-1, IL-1ß, TNF-α, Bax, and Bcl-2 were determined by Western Blot. Neutrophils, microglia were measured by immunohistochemistry staining, neuronal apoptosis was measured by TUNEL and NeuN co-stained. Administration of MPSS post-ICH significantly reduced permeability of the BBB and brain oedema and upregulated expression of ZO-1 and Occludin. MPSS inhibited inflammatory responses, including reducing proinflammatory cytokines (IL-1ß, TNF-α), suppressing infiltration of neutrophils and activation of microglia. This was accompanied by attenuated activation of the TLR4/NF-κB signalling pathway. In addition, MPSS reduced neuronal apoptosis through increasing Bcl-2 expression and reducing Bax expression. MPSS suppressed inflammatory responses, attenuated disruption of the BBB and reduced neuronal apoptosis, contributing to reduction of secondary brain injury after ICH. These results suggest that MPSS may be a potential therapy for ICH.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Blood-Brain Barrier/drug effects , Inflammation/drug therapy , Intracranial Hemorrhages/drug therapy , Methylprednisolone Hemisuccinate/pharmacology , Neuroprotective Agents/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Astrocytes/drug effects , Astrocytes/immunology , Astrocytes/pathology , Blood-Brain Barrier/immunology , Blood-Brain Barrier/pathology , Brain Edema/drug therapy , Brain Edema/immunology , Brain Edema/pathology , Cytokines/metabolism , Disease Models, Animal , Inflammation/metabolism , Inflammation/pathology , Intracranial Hemorrhages/immunology , Intracranial Hemorrhages/pathology , Male , Mice, Inbred C57BL , Microglia/drug effects , Microglia/immunology , Microglia/pathology , Neurons/drug effects , Neurons/immunology , Neurons/pathology , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/pathology , Random Allocation
6.
Arterioscler Thromb Vasc Biol ; 36(8): 1517-24, 2016 08.
Article in English | MEDLINE | ID: mdl-27283740

ABSTRACT

OBJECTIVE: Fetal/neonatal alloimmune thrombocytopenia is a severe bleeding disorder, which can result in intracranial hemorrhage (ICH), leading to death or neurological sequelae. In whites, maternal anti-human platelet antigen-1a (HPA-1a) antibodies are responsible for the majority of cases. No predictive factors for ICH are available to guide prophylactic treatment during pregnancy. In this study, we investigated antibodies from mothers with ICH-positive fetal/neonatal alloimmune thrombocytopenia and with ICH-negative fetal/neonatal alloimmune thrombocytopenia to identify serological and functional differences between the groups. APPROACH AND RESULTS: In an antigen capture assay, we observed a stronger binding of +ICH antibodies to endothelial cell (EC)-derived αvß3. By absorption experiments, we subsequently identified anti-HPA-1a antibodies of anti-αvß3 specificity in the +ICH but not in the -ICH cohort. Only the anti-αvß3 subtype, but not the anti-ß3 subtype, induced EC apoptosis of HPA-1a-positive ECs by caspase-3/7 activation, and mediated by reactive oxygen species. In addition, only the anti-αvß3 subtype, but not the anti-ß3 subtype, interfered with EC adhesion to vitronectin and with EC tube formation. CONCLUSIONS: We conclude that the composition of the anti-HPA-1a antibody subtype(s) of the mother may determine whether ICH occurs. Analysis of anti-HPA-1a antibodies of the anti-αvß3 subtype in maternal serum has potential in the diagnostic prediction of ICH development and may allow for modification of prophylactic treatment in fetal/neonatal alloimmune thrombocytopenia.


Subject(s)
Autoantibodies/immunology , Endothelial Cells/immunology , Integrin alphaVbeta3/immunology , Intracranial Hemorrhages/immunology , Thrombocytopenia, Neonatal Alloimmune/immunology , Animals , Antibody Specificity , Antigens, Human Platelet/immunology , Antigens, Human Platelet/metabolism , Apoptosis , Autoantibodies/metabolism , CHO Cells , Case-Control Studies , Caspase 3/metabolism , Caspase 7/metabolism , Cell Adhesion , Cricetulus , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Gestational Age , Human Umbilical Vein Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , Infant, Newborn , Integrin alphaVbeta3/genetics , Integrin alphaVbeta3/metabolism , Integrin beta3 , Intracranial Hemorrhages/metabolism , Intracranial Hemorrhages/pathology , Male , Maternal-Fetal Exchange , Neovascularization, Physiologic , Pregnancy , Reactive Oxygen Species/metabolism , Thrombocytopenia, Neonatal Alloimmune/metabolism , Thrombocytopenia, Neonatal Alloimmune/pathology , Transfection
7.
Brain Behav Immun ; 58: 118-129, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27261088

ABSTRACT

Excessive inflammatory responses are involved in secondary brain injury during germinal matrix hemorrhage (GMH). The process of microglial polarization to the pro-inflammatory M1 or anti-inflammatory M2 phenotypes is considered to occur in a major immunomodulatory manner during brain inflammation. We previously found that cannabinoid receptor-2 (CB2R) stimulation attenuated microglial accumulation and brain injury following experimental GMH. However, whether CB2R has effects on microglial polarization after GMH remains unclear. Herein, we investigated the effects of CB2R stimulation on neuroinflammation after experimental GMH and the potential mechanisms that mediate M1/M2 microglial phenotype regulation. The results indicated that during the GMH acute phase, microglia primarily polarized to the M1 phenotype and induced an overwhelming release of pro-inflammatory cytokines. However, JWH133, a selective CB2R agonist, significantly prevented the pro-inflammatory cytokine release while promoting an M1 to M2 phenotype transformation in microglia, resulting in an increased anti-inflammatory cytokine release. Moreover, in thrombin-induced rat primary microglial cells, JWH133 reduced the pro-inflammatory cytokine levels and M1 phenotype by enhancing the acquisition of the M2 phenotype. Additionally, JWH133 facilitated synthesis of cyclic AMP (cAMP) and its downstream effectors, phosphorylated cAMP-dependent protein kinase (p-PKA) and exchange protein activated by cyclic-AMP 1 (Epac1). The promoting effects of JWH133 on M2 polarization were attenuated with a specific PKA inhibitor but not with an Epac inhibitor, indicating that the cAMP/PKA signaling pathway was involved in the JWH133 effects. This is the first study to propose that promotion of microglial M2 polarization through the cAMP/PKA pathway participates in the CB2R-mediated anti-inflammatory effects after GMH induction. The results will help to further understand the mechanisms that underlie neuroprotection by CB2R in GMH and promote clinical translational research for CB2R agonists.


Subject(s)
Encephalitis/immunology , Intracranial Hemorrhages/immunology , Microglia/immunology , Receptor, Cannabinoid, CB2/metabolism , Animals , Cell Polarity , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytokines/metabolism , Disease Models, Animal , Encephalitis/metabolism , Intracranial Hemorrhages/metabolism , Male , Microglia/metabolism , Rats, Sprague-Dawley , Signal Transduction
8.
Biochem Biophys Res Commun ; 468(1-2): 228-33, 2015.
Article in English | MEDLINE | ID: mdl-26523514

ABSTRACT

During pregnancy, both ischemic reperfusion and bacterial agent LPS are known risk factors for fetal brain damage. However, there is a lack of evidence to explain whether vaginal LPS affects the fetus response to ischemic reperfusion. Here we reported that there was more than 2 folds higher vulnerability of fetal brain hemorrhage response to ischemic reperfusion when mother mouse was treated with vaginal LPS. As our previously reported, ischemic reperfusion induces P53-dependent fetal brain damage was based on a molecular mechanism: the transcriptional pattern was changed from HIF-1alpha-dependent to P53-dependent immediately. In the present work, only with vaginal LPS precondition, phosphorylation of activated transcriptional factor (ATF) 2 at Thr71 appeared in response to ischemic reperfusion. Moreover, this phosphorylation was completely blocked by pre-treatment with a P53 inhibitor, pifithrin-α. We concluded that vaginal LPS precondition trigged the p53-dependent phosphorylation of ATF2 in response to ischemic reperfusion, which played an important role of increasing vulnerability to hemorrhage in fetus.


Subject(s)
Brain/pathology , Fetal Diseases/etiology , Fetus/pathology , Intracranial Hemorrhages/etiology , Lipopolysaccharides/immunology , Reperfusion Injury/complications , Vagina/immunology , Activating Transcription Factor 2/analysis , Activating Transcription Factor 2/immunology , Animals , Brain/immunology , Brain/metabolism , Female , Fetal Diseases/genetics , Fetal Diseases/immunology , Fetal Diseases/pathology , Fetus/immunology , Fetus/metabolism , Inflammation/complications , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Intracranial Hemorrhages/genetics , Intracranial Hemorrhages/immunology , Intracranial Hemorrhages/pathology , Mice , Mice, Inbred C57BL , Pregnancy , Reperfusion Injury/genetics , Reperfusion Injury/immunology , Reperfusion Injury/pathology , Transcriptional Activation , Tumor Necrosis Factor-alpha/immunology , Tumor Suppressor Protein p53/immunology
9.
J Clin Invest ; 125(4): 1545-56, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25774504

ABSTRACT

Fetal and neonatal alloimmune thrombocytopenia (FNAIT) is a life-threatening disease in which intracranial hemorrhage (ICH) is the major risk. Although thrombocytopenia, which is caused by maternal antibodies against ß3 integrin and occasionally by maternal antibodies against other platelet antigens, such as glycoprotein GPIbα, has long been assumed to be the cause of bleeding, the mechanism of ICH has not been adequately explored. Utilizing murine models of FNAIT and a high-frequency ultrasound imaging system, we found that ICH only occurred in fetuses and neonates with anti-ß3 integrin-mediated, but not anti-GPIbα-mediated, FNAIT, despite similar thrombocytopenia in both groups. Only anti-ß3 integrin-mediated FNAIT reduced brain and retina vessel density, impaired angiogenic signaling, and increased endothelial cell apoptosis, all of which were abrogated by maternal administration of intravenous immunoglobulin (IVIG). ICH and impairment of retinal angiogenesis were further reproduced in neonates by injection of anti-ß3 integrin, but not anti-GPIbα antisera. Utilizing cultured human endothelial cells, we found that cell proliferation, network formation, and AKT phosphorylation were inhibited only by murine anti-ß3 integrin antisera and human anti-HPA-1a IgG purified from mothers with FNAIT children. Our data suggest that fetal hemostasis is distinct and that impairment of angiogenesis rather than thrombocytopenia likely causes FNAIT-associated ICH. Additionally, our results indicate that maternal IVIG therapy can effectively prevent this devastating disorder.


Subject(s)
Antigens, Human Platelet/immunology , Autoantigens/immunology , Blood Platelets/immunology , Immunity, Maternally-Acquired , Immunoglobulin G/immunology , Immunoglobulins, Intravenous/therapeutic use , Integrin beta3/immunology , Intracranial Hemorrhages/etiology , Neovascularization, Pathologic/etiology , Thrombocytopenia, Neonatal Alloimmune/immunology , Animals , Antibody Specificity , Apoptosis , Brain/blood supply , Brain/embryology , Disease Models, Animal , Female , Fetal Blood/immunology , Human Umbilical Vein Endothelial Cells , Humans , Immune Sera/toxicity , Integrin beta3/genetics , Intracranial Hemorrhages/embryology , Intracranial Hemorrhages/immunology , Intracranial Hemorrhages/physiopathology , Male , Maternal-Fetal Exchange , Mice , Mice, Knockout , Neovascularization, Physiologic/immunology , Platelet Glycoprotein GPIb-IX Complex/genetics , Platelet Glycoprotein GPIb-IX Complex/immunology , Pregnancy , Proto-Oncogene Proteins c-akt/physiology , Retinal Vessels/embryology , Retinal Vessels/pathology , Thrombocytopenia, Neonatal Alloimmune/embryology , Thrombocytopenia, Neonatal Alloimmune/prevention & control
10.
Eur J Pediatr ; 173(2): 163-72, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24390128

ABSTRACT

Immune thrombocytopenia (ITP) is a disease affecting both children and adults. It is defined as acquired isolated thrombocytopenia caused by the autoimmune production of anti-platelet antibodies. Childhood ITP most frequently occurs in young children who have been previously well, although a viral respiratory tract infection often precedes thrombocytopenia. A benign and self-limiting course is common, but major bleeding complications such as intracranial haemorrhage may occur. Yet one cannot predict which child will have a prolonged course of thrombocytopenia and who will develop an intracranial haemorrhage. In children without atypical characteristics, only minimal diagnostic investigations are needed, and most paediatric ITP patients do not need platelet-enhancing therapy even though various treatment options are available. A "watch and wait" strategy should be considered in paediatric patients with mild disease. Steroids, intravenous immunoglobulin G or anti-D immunoglobulin are the current first-line therapeutic measures for children at risk for severe bleeding. When life-threatening bleeding occurs, a combination of therapies is needed. In this review, we summarise the current knowledge on primary ITP in children and adolescents.


Subject(s)
Purpura, Thrombocytopenic, Idiopathic/diagnosis , Adolescent , Adrenal Cortex Hormones/therapeutic use , Autoantibodies/blood , Blood Platelets/immunology , Child , Child, Preschool , Humans , Immunization, Passive , Immunoglobulin G/therapeutic use , Intracranial Hemorrhages/etiology , Intracranial Hemorrhages/immunology , Intracranial Hemorrhages/prevention & control , Isoantibodies/therapeutic use , Prognosis , Purpura, Thrombocytopenic, Idiopathic/complications , Purpura, Thrombocytopenic, Idiopathic/immunology , Purpura, Thrombocytopenic, Idiopathic/therapy , Respiratory Tract Infections/complications , Rho(D) Immune Globulin , Risk Factors , Watchful Waiting
11.
J Neurosurg Sci ; 57(1): 55-62, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23584220

ABSTRACT

AIM: Recent experimental studies have suggested that chemokines, a subclass of chemoattractant cytokines which play an important role in regulating leukocyte migration and intercellular communication, participate in brain responses of traumatic injury. Fractalkine (CX3CL1) is a peculiar chemokine, the only one with a CX3C motif, existing both as a soluble and a membrane-anchored molecule. In the brain, Fractalkine has been suggested to have a role in neuroprotection under experimental conditions of brain injury. METHODS: Eighteen human brain samples were obtained during surgery of decompressive craniotomy for severe traumatic brain injury (TBI) or after spontaneous intracranial haemorrhage (ICH). Five normal brain samples were obtained during surgery for unruptured intracranial aneurysms (standard gyrectomy). Immunohistochemistry of formalin fixed and paraffin embedded tissues was performed in order to verify the expression of fractalkine and its receptor (CX3CR1). The values of chemokine and receptor expression were correlated with the clinical parameters of the patients. RESULTS: The chemokine fractalkine was significantly upregulated in the neural compartment after brain injury, compared to normal brain samples. Intensity scores were significantly higher when the interval between injury and surgery was >5 h, (P=0.015). In the glial compartment, Fractalkine expression was significantly associated with less severe clinical conditions and lower intracranial pressure at surgery (P=0.014). Expression of the receptor CX3CR1 was detected, at low intensity, on both glial and neurons. Higher expression in neurons was associated with better clinical conditions (Glasgow score) of patients at admission (P=0.037). CONCLUSION: The results of this study highlights for the first time that fractalkine and its receptor CX3CR1 are expressed in the human brain after TBI and ICH and may be involved in the limitation of tissue damage.


Subject(s)
Brain Injuries/metabolism , Chemokine CX3CL1/metabolism , Intracranial Hemorrhages/metabolism , Receptors, Chemokine/metabolism , Severity of Illness Index , Adult , Aged , Brain Injuries/immunology , Brain Injuries/pathology , CX3C Chemokine Receptor 1 , Chemokine CX3CL1/immunology , Female , Humans , Immunohistochemistry , Intracranial Hemorrhages/immunology , Intracranial Hemorrhages/pathology , Male , Middle Aged , Neuroglia/immunology , Neuroglia/metabolism , Receptors, Chemokine/immunology , Time Factors , Young Adult
12.
Neuropharmacology ; 67: 267-71, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23219658

ABSTRACT

Ischemic and hemorrhagic strokes have different etiologies, but share some pathogenic mechanisms, including a pro-neurotoxic effect of endogenous tissue plasminogen activator (tPA) via N-methyl-d-Aspartate (NMDA) receptors. Thus, in a model of intracerebral hemorrhage in rats, we investigated the therapeutic value of a strategy of immunotherapy (αATD-GluN1 antibody) preventing the interaction of tPA with NMDA receptors. We found that a single intravenous injection of αATD-GluN1 reduced brain edema, neuronal death, microglial activation and functional deficits following intracerebral hemorrhage, without affecting the hematoma volume.


Subject(s)
Immunotherapy/methods , Intracranial Hemorrhages/immunology , Intracranial Hemorrhages/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Stroke/immunology , Stroke/metabolism , Tissue Plasminogen Activator/antagonists & inhibitors , Animals , Antibodies, Monoclonal, Murine-Derived/administration & dosage , Intracranial Hemorrhages/therapy , Male , Mice , Random Allocation , Rats , Rats, Sprague-Dawley , Stroke/therapy , Tissue Plasminogen Activator/physiology , Treatment Outcome
13.
Aust N Z J Obstet Gynaecol ; 52(4): 321-6, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22510050

ABSTRACT

BACKGROUND: Fetal and neonatal alloimmune thrombocytopenia (FNAIT) is the most common cause of severe perinatal thrombocytopenia, arising from the transplacental passage of maternal antibodies directed at paternally inherited antigens on fetal platelets. AIM: To review the occurrence, management and perinatal outcomes of pregnancies complicated by FNAIT from a single tertiary obstetric hospital in Western Australia. MATERIALS AND METHODS: The study was conducted as a retrospective review of cases with prenatally recognised FNAIT between 2001 and 2011, with the treatment modalities and outcomes analysed. RESULTS: Over the 10-year period, 20 cases of clinically significant FNAIT in 13 women were managed at our centre. Three cases were complicated by antenatal intracranial haemorrhage (15%), and in all 3, this was the presenting feature leading to diagnosis. In 17/20 (85%) cases, anti-HPA 1a was the responsible antibody, with the remainder being anti-HPA 5b. In 16/17 cases with pre-pregnancy recognition, intravenous gammaglobulin (IVGG) was administered antenatally (gestation at commencement ranging from 13 to 26 weeks) with adjuvant prednisolone in three cases. Postnatal treatment (IVGG or platelet transfusion) was provided in 4/16 cases. There was no intracranial haemorrhage or demise in any case receiving prenatal therapy. CONCLUSIONS: FNAIT is a rare and serious condition. In our small single-centre study, there was variability in the therapeutic strategies, although IVGG was central to all prenatally managed pregnancies. None of the treated pregnancies was complicated by intracranial haemorrhage or fetal death. There is a need for ongoing refinement of FNAIT management protocols, both in the prenatal and in the postnatal period.


Subject(s)
Fetal Diseases/therapy , Immunoglobulins, Intravenous/therapeutic use , Intracranial Hemorrhages/etiology , Prednisolone/therapeutic use , Thrombocytopenia, Neonatal Alloimmune/therapy , Adult , Female , Fetal Diseases/diagnosis , Fetal Diseases/immunology , Fetus/immunology , Humans , Infant, Newborn , Intracranial Hemorrhages/immunology , Intracranial Hemorrhages/therapy , Platelet Count , Platelet Transfusion/statistics & numerical data , Pregnancy , Retrospective Studies , Tertiary Care Centers , Thrombocytopenia/complications , Thrombocytopenia/diagnosis , Thrombocytopenia/immunology , Thrombocytopenia/therapy , Thrombocytopenia, Neonatal Alloimmune/diagnosis , Western Australia
14.
Aust N Z J Obstet Gynaecol ; 51(3): 191-8, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21631435

ABSTRACT

Fetomaternal or neonatal alloimmune thrombocytopenia (NAIT) is a rare but serious condition associated with significant fetal and neonatal morbidity and mortality. The most useful predictor of severe disease is a history of a sibling with an antenatal intracranial haemorrhage. However, NAIT can occur during the first pregnancy and may not be diagnosed until the neonatal period. Antenatal treatment options include maternal intravenous immunoglobulin (IVIG) and corticosteroid treatment, fetal blood sampling (FBS) and intrauterine platelet transfusion (IUT) and early delivery. FBS (with or without IUT) can be used to direct and monitor response to therapy, and to inform mode and timing of delivery. However, this procedure is associated with significant risks, including fetal death, and is generally now reserved for high-risk pregnancies. This review highlights the current understanding of the epidemiology and pathophysiology of NAIT and summarises current approaches to investigation and management. It also introduces the newly established Australian NAIT registry. Owing to the relative rarity of NAIT, accruing sufficient patient numbers for studies and clinical trials at an institutional level is difficult. This national registry will provide an opportunity to collect valuable information and inform future research on this condition.


Subject(s)
Registries , Thrombocytopenia, Neonatal Alloimmune/physiopathology , Thrombocytopenia, Neonatal Alloimmune/therapy , Adrenal Cortex Hormones/therapeutic use , Antigens, Human Platelet/genetics , Antigens, Human Platelet/immunology , Australia , Blood Specimen Collection/adverse effects , Blood Transfusion, Intrauterine , Female , Fetal Blood , Fetal Mortality , Humans , Immunoglobulins, Intravenous/therapeutic use , Infant Mortality , Infant, Newborn , Intracranial Hemorrhages/immunology , Intracranial Hemorrhages/therapy , Platelet Transfusion , Pregnancy , Pregnancy, High-Risk , Prenatal Care , Thrombocytopenia, Neonatal Alloimmune/diagnosis
15.
Pediatr Res ; 70(1): 90-5, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21659962

ABSTRACT

Intraventricular hemorrhage (IVH) is a significant morbidity seen in very LBW infants. Genes related to the inflammation, infection, complement, or coagulation pathways have been implicated as risk factors for IVH. We examined 10 candidate genes for associations with IVH in 271 preterm infants (64 with IVH grades I-IV and 207 without IVH) weighing <1500 g. The heterozygous genotype OR = 8.1, CI = 2.5-26.0, p = 4 × 10(-4)) and the A allele (OR = 7.3, CI = 2.4-22.5, p = 1 × 10(-4)) of the coagulation factor V (FV) Leiden mutation (rs6025) were associated with an increased risk of developing IVH grade I or II but not grade III or IV after correction for multiple testing with Bonferroni. Lack of association in the severe grades of IVH may be a result of lack of power to detect an effect given the small sample size (n = 8). However, this result is consistent with previous research that demonstrates that the heterozygous genotype of the FV mutation is associated with increased risk for the development of IVH but a decreased risk for the progression or extension to more severe grades of IVH.


Subject(s)
Blood Coagulation/genetics , Complement System Proteins/genetics , Infant, Low Birth Weight , Infant, Premature , Inflammation/genetics , Intracranial Hemorrhages/genetics , Polymorphism, Single Nucleotide , Cytokines/genetics , Estrogen Receptor alpha/genetics , Factor V/genetics , Female , Gene Frequency , Genetic Association Studies , Genetic Predisposition to Disease , Gestational Age , Heterozygote , Humans , Infant, Newborn , Inflammation/blood , Inflammation/immunology , Inflammation Mediators/metabolism , Integrin beta3/genetics , Intracranial Hemorrhages/blood , Intracranial Hemorrhages/immunology , Iowa , Linkage Disequilibrium , Logistic Models , Male , Odds Ratio , Phenotype , Reproducibility of Results , Risk Assessment , Risk Factors , Severity of Illness Index
16.
BMJ Case Rep ; 20112011 Sep 04.
Article in English | MEDLINE | ID: mdl-22679192

ABSTRACT

Neonatal alloimmune thrombocytopenia is a rare (1/1000-5000 births) life-threatening disorder, caused by fetomaternal incompatibility for a fetal human platelet alloantigen inherited from the father, with production of maternal alloantibodies against fetal platelets, leading to severe thrombocytopenia and potential bleeding. Intracranial haemorrhage is the most feared complication. This report presents the case of a term newborn infant, born from caesarean section after a normal pregnancy, presenting signs of skin bleeding with different ages. Obstetric history included a previous spontaneous abortion after amniocentesis. Severe thrombocytopenia (4×10(9)/l platelets) was found and brain ultrasound showed multiple intracranial haemorrhages. Human platelet antigen (HPA) phenotyping showed maternal negative HPA-1a and paternal positive HPA-1a platelets. Strongly positive anti-HPA-1a and weakly positive anti-human leukocyte antigen class I alloantibodies were found in the mother. Multiple platelet transfusions, intravenous immunoglobulin and corticosteroid were given but favourable response was accomplished only after a compatible platelet transfusion. Brain MRI showed multiple subacute and chronic haemorrhages.


Subject(s)
Intracranial Hemorrhages/etiology , Intracranial Hemorrhages/immunology , Thrombocytopenia, Neonatal Alloimmune/diagnosis , Female , Glucocorticoids/therapeutic use , Humans , Infant, Newborn , Intracranial Hemorrhages/therapy , Magnetic Resonance Imaging , Methylprednisolone/therapeutic use , Platelet Transfusion , Thrombocytopenia, Neonatal Alloimmune/therapy , Ultrasonography, Prenatal
17.
CNS Neurol Disord Drug Targets ; 8(1): 50-64, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19275636

ABSTRACT

Alzheimer's disease (AD) is a progressive, neurodegenerative disorder that results in severe cognitive decline. Amyloid plaques are a principal pathology found in AD and are composed of aggregated amyloid-beta (Abeta) peptides. According to the amyloid hypothesis, Abeta peptides initiate the other pathologies characteristic for AD including cognitive deficits. Immunotherapy against Abeta is a potential therapeutic for the treatment of humans with AD. While anti-Abeta immunotherapy has been shown to reduce amyloid burden in both mouse models and in humans, immunotherapy also exacerbates vascular pathologies. Cerebral amyloid angiopathy (CAA), that is, the accumulation of amyloid in the cerebrovasculature, is increased with immunotherapy in humans with AD and in mouse models of amyloid deposition. CAA persists in the brains of clinical trial patients that show removal of parenchymal amyloid. Mouse model studies also show that immunotherapy results in multiple small bleeds in the brain, termed microhemorrhages. The neurovascular unit is a term used to describe the cerebrovasculature and its associated cells-astrocytes, neurons, pericytes and microglia. CAA affects brain perfusion and there is now evidence that the neurovascular unit is affected in AD when CAA is present. Understanding the type of damage to the neurovascular unit caused by CAA in AD and the underlying cause of microhemorrhage after immunotherapy is essential to the success of therapeutic vaccines as a treatment for AD.


Subject(s)
Alzheimer Disease/therapy , Amyloid beta-Peptides/metabolism , Cerebral Amyloid Angiopathy/complications , Immunotherapy/adverse effects , Intracranial Hemorrhages/etiology , Alzheimer Disease/complications , Alzheimer Disease/immunology , Alzheimer Disease/metabolism , Amyloid beta-Peptides/immunology , Animals , Antibodies/immunology , Antibodies/metabolism , Cerebral Amyloid Angiopathy/immunology , Humans , Immunotherapy/methods , Intracranial Hemorrhages/immunology , Mice , Mice, Transgenic
18.
Bull Exp Biol Med ; 148(3): 530-2, 2009 Sep.
Article in English, Russian | MEDLINE | ID: mdl-20396732

ABSTRACT

Ultralow doses of antibodies to S-100 protein increased rat survival, reduced neurological deficit, eliminated myorelaxation, and improved movement coordination and cognitive functions in rats with experimental hemorrhagic stroke; the efficiency of the preparation was not inferior to that of nimodipine. In contrast to nimodipine, ultralow doses of antibodies to S-100 protein exhibited pronounced anxiolytic properties.


Subject(s)
Antibodies/therapeutic use , Intracranial Hemorrhages/drug therapy , S100 Proteins/immunology , Animals , Antibodies/immunology , Intracranial Hemorrhages/immunology , Male , Nimodipine/therapeutic use , Rats , Vasodilator Agents/therapeutic use
19.
Iran J Allergy Asthma Immunol ; 7(3): 181-4, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18780954

ABSTRACT

Immunodeficiency and autoimmune disease may occur concomitantly in the same individual. Some of the immunodeficiency syndromes, especially humoral defects are associated with autoimmune disorders. Hematological manifestations such as thrombocytopenia and hemolytic anemia are the most common presentations. Persistent antigen stimulation due to an inherent defect in the ability of the immune system to eradicate pathogens is the primary cause leading to autoimmunity in patients with primary immunodeficiency states.We describe a 10 year old Iranian girl with chronic granulomatous disease -the autosomal recessive type with mutation of NCF1 gene P47- associated with selective IgA deficiency, refractory immune thrombocytopenia that showed an excellent response to Rituximab (Anti-CD20 monoclonal antibody).Patients with primary immunodeficiencies may have variable autoimmune manifestations. So for early detection and appropriate treatment, autoimmune diseases should always be suspected in such patients.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Granulomatous Disease, Chronic/drug therapy , IgA Deficiency/drug therapy , IgA Deficiency/immunology , Immunologic Factors/therapeutic use , Purpura, Thrombocytopenic, Idiopathic/drug therapy , Antibodies, Monoclonal, Murine-Derived , Child , DNA Mutational Analysis , Female , Genes, Recessive , Granulomatous Disease, Chronic/complications , Granulomatous Disease, Chronic/genetics , Granulomatous Disease, Chronic/immunology , Humans , IgA Deficiency/complications , IgA Deficiency/genetics , Immunoglobulins/blood , Intracranial Hemorrhages/immunology , Iran , Mutation , NADPH Oxidases/genetics , Platelet Count , Purpura, Thrombocytopenic, Idiopathic/etiology , Purpura, Thrombocytopenic, Idiopathic/immunology , Respiratory Tract Infections/immunology , Rituximab
20.
Phytother Res ; 22(8): 1083-6, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18570265

ABSTRACT

Multiple sclerosis is an inflammatory and demyelinating disease of the central nervous system which mainly affects young adults. To overcome wide spectrum troublesome symptoms of multiple sclerosis which affects the quality of life both in patients and their families, new drugs and remedies have been examined and offered. The preclinical beneficial effects of different medicines have mostly been examined in an animal model of multiple sclerosis called experimental allergic encephalomyelitis (EAE). In this study we have tested a traditionally used natural (herbal-marine) product called MS(14) in EAE mice. EAE mice were fed with MS(14) containing diet (30%) on the immunization day and monitored for 20 days. The results show that while clinical scores and therefore severity of the disease was progressive in normal-fed EAE mice, the disease was slowed down in MS(14)-fed EAE mice. Moreover, while there were moderate to severe neuropathological changes in normal fed mice, milder changes were seen in MS(14) fed mice.


Subject(s)
Biological Products/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Administration, Oral , Animal Feed , Animals , Apium/chemistry , Biological Products/chemistry , Brain/drug effects , Brain/pathology , Demyelinating Diseases/drug therapy , Demyelinating Diseases/immunology , Demyelinating Diseases/pathology , Disease Models, Animal , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Female , Hypericum/physiology , Intracranial Hemorrhages/drug therapy , Intracranial Hemorrhages/immunology , Intracranial Hemorrhages/pathology , Iran , Mice , Necrosis , Penaeidae/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...